Sie sind auf Seite 1von 22

Article https://doi.org/10.

1038/s41586-018-0832-5

GABAA receptor signalling mechanisms


revealed by structural pharmacology
Simonas Masiulis1*, Rooma Desai2, Tomasz Uchański3,4, Itziar Serna Martin5, Duncan Laverty1, Dimple Karia5,
Tomas Malinauskas5, Jasenko Zivanov1, Els Pardon3,4, Abhay Kotecha6, Jan Steyaert3,4, Keith W. Miller2* & A. Radu Aricescu1,5*

Type-A γ-aminobutyric (GABAA) receptors are ligand-gated chloride channels with a very rich pharmacology. Some of
their modulators, including benzodiazepines and general anaesthetics, are among the most successful drugs in clinical
use and are common substances of abuse. Without reliable structural data, the mechanistic basis for the pharmacological
modulation of GABAA receptors remains largely unknown. Here we report several high-resolution cryo-electron
microscopy structures in which the full-length human α1β3γ2L GABAA receptor in lipid nanodiscs is bound to the
channel-blocker picrotoxin, the competitive antagonist bicuculline, the agonist GABA (γ-aminobutyric acid), and the
classical benzodiazepines alprazolam and diazepam. We describe the binding modes and mechanistic effects of these
ligands, the closed and desensitized states of the GABAA receptor gating cycle, and the basis for allosteric coupling
between the extracellular, agonist-binding region and the transmembrane, pore-forming region. This work provides a
structural framework in which to integrate previous physiology and pharmacology research and a rational basis for the
development of GABAA receptor modulators.

In vertebrates, GABAA receptors mediate both phasic and tonic neuronal (ALP, also known by the trade name Xanax) act specifically through
inhibition in the adult central nervous system1–3. Their dysfunction GABAA receptors containing α1, α2, α3 or α5, but not α4 or α6 sub-
leads to channelopathies associated with epilepsy, insomnia, anxiety unit types22,23. The basis for this specificity is not known. The plant
and chronic pain4. GABAA receptors are among the most important alkaloid PTX, which is one of the most widely used GABAA receptor
human drug targets owing to their many allosteric sites, which bind antagonists24, is thought to act as a channel blocker. However, its com-
compounds with anticonvulsant, anti-anxiety, analgesic, sedative and petitive antagonistic properties suggest that additional binding sites
anaesthetic properties5,6. Some of these—such as benzodiazepines might exist25,26. Another broadly used GABAA receptor antagonist,
(BZDs), which are typically positive allosteric modulators—entered BCC, is thought to act competitively, although its preference for bind-
clinical use decades before the identity of their receptors was known7,8, ing resting over desensitized receptors suggests that it might also act
and were crucial for the isolation9 and subsequent cloning10 of GABAA allosterically27. How reagents such as PTX and BCC actually bind, as
receptors. Other GABAA receptor ligands are important research tools, well as how they work, is not yet known.
including the antagonists picrotoxin (PTX) and bicuculline (BCC)5,6. To address these questions, here we present five structures of the
The binding modes and conformational effect of most GABAA human synaptic α1β3γ2L GABAA receptor in complex with PTX, PTX
receptor allosteric modulators are unknown. Docking attempts have and GABA (PTX/GABA), BCC, DZP and GABA (DZP/GABA), and
had to rely on models based on the structures of homologous pro- ALP and GABA (ALP/GABA). We establish the binding modes and
teins, including the Caenorhabditis elegans glutamate-gated chloride structural effect of these ligands and explain the molecular basis for
channel α (GluCl)11, the Torpedo nicotinic acetylcholine receptor12 or their function. To obtain structures in which GABAA receptors can
the human homopentameric β3 GABAA receptor13. Crystallographic be observed in physiologically relevant conformations, we used a full-
and single-particle cryo-electron microscopy (cryo-EM) studies have length receptor variant from a thoroughly characterized cell line28 and
revealed the interactions of GABAA-receptor-derived constructs with reconstituted it in a lipid bilayer29. Our results lay the foundation for
neurosteroids14–16, the agonist GABA17–19, and the BZD-site ligands understanding the fundamental principles of the action of small mol-
bretazenil (BRZ)20 and flumazenil (FLZ)18,20. However, owing to the use ecules on heteromeric synaptic GABAA receptors.
of engineered receptors, some of which are structurally damaged by the
presence of detergents, the interpretability of these models is limited. Picrotoxin and GABA binding modes
Functional studies on small-molecule GABAA receptor modulators We first solved the structure of the α1β3γ2L receptor in complex with
have also raised numerous questions. We will list here just a few examples. PTX (800 μM) to a nominal resolution of 3.1 Å (Fig. 1a, Extended Data
It is not known why the two agonist (GABA) binding sites, which Fig. 1a–f, Extended Data Table 1, Supplementary Video 1). PTX is an
should be structurally identical, are not functionally equivalent21. The equimolar mix of two highly similar compounds, picrotin and picro-
mechanism of action of BZDs is also unclear, despite their widespread toxinin. Our structure illustrates picrotoxinin, which is known to be the
use as sedatives and anxiolytics. Moreover—unlike newer compounds more active compound, fully sequestered in the channel pore between
such as bretazenil and flumazenil—classical BZDs including diaze- the M2 2ʹ and 9ʹ rings (Fig. 1b, c, Extended Data Fig. 1g, Supplementary
pam (DZP, also known by the trade name Valium) and alprazolam Video 2). This agrees both with mutagenesis and electrophysiology

1
MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK. 2Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard
Medical School, Boston, MA, USA. 3Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium. 4VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium. 5Division of
Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK. 6Materials and Structural Analysis, Thermo Fisher Scientific, Eindhoven, The Netherlands.
*e-mail: simonasm@mrc-lmb.cam.ac.uk; kwmiller@mgh.harvard.edu; radu@mrc-lmb.cam.ac.uk

N A T U R E | www.nature.com/nature
© 2018 Springer Nature Limited. All rights reserved.
RESEARCH Article

a α1-D γ2-C b c
β3-E M2 M2 M2
N-linked γ2-C
Mb38 Loop-C β3-B M2
glycans
α1-A L274
L259, 9′
L274, 9′
M2 T271
L264, 9′
β3-B 72°
T271, 6′ 90°
T256, 6′ M2
β3-E Out
T261, 6′ T261

90° α1-D
L259
A252, 2′

TMD
PTX M2
T256
Loop-C γ2-C V257, 2′
α1-D S267, 2′ L264 M2
In α1-A β3-E

ICD
PIP2 β3-B γ2-C α1-D

Fig. 1 | Structure of the α1β3γ2L GABAA receptor in complex with down (c) views of PTX (carbon atoms in pink, oxygen atoms in red) bound
picrotoxin. a, Cryo-EM map of the PTX-bound α1β3γ2L GABAA receptor to the channel pore, with the amino acid side chains lining the site shown
viewed from the extracellular space (left) and parallel to the membrane as sticks. Dashed lines indicate hydrogen bonds.
plane (right). The PTX-binding site is boxed. b, c, Side-on (b) and top-

results that implicate the 2ʹ, 6ʹ and 9′ M2 residues in PTX binding30–32, sites are located at the two β3+/α1− interfaces (the principal (+) and
and with suggestions that PTX becomes ‘trapped’ in closed/rest- complementary (−) annotation of subunit faces is used here), capped
ing channels of GABAA and Gly receptors upon agonist wash-off33,34. by the extracellular loops-C of the β subunits18,19,29. Similar to the
The hydrophobic isoprenyl moiety is surrounded by the 9ʹ Leu ring, Torpedo nAChR α subunits35, in the agonist-free GABAA receptor
whereas the exocyclic oxygen atoms in the main PTX body form puta- the β3 loops-C adopt ‘open’ (outwards-projecting) conformations,
tive hydrogen bonds with the 6ʹ ring (Fig. 1b, c). In picrotin, the isoprenyl whereas in the PTX/GABA-bound state they are ‘closed’ (Extended
group is replaced by a polar tertiary alcohol; this is not compatible Data Fig. 2d). Strong electron microscopy densities are present in the
with 9′ Leu ring interactions, which explains why picrotin is less active. two orthosteric ligand-binding sites, enabling unambiguous model-
We next determined a GABAA receptor structure in complex with ling of GABA molecules in the ‘aromatic boxes’ formed by β3Tyr157,
PTX (800 μM) and the neurotransmitter GABA (5 μM) to a nominal β3Phe200, β3Tyr205 and α1Phe65 (Fig. 2b, Extended Data Fig. 2e,
resolution of 3.04 Å (Fig. 2a, Extended Data Fig. 2a–c, Extended Data Supplementary Video 4). The GABA amino group engages in a cation–π
Table 1, Supplementary Video 3). The orthosteric GABA-binding interaction with β3Tyr205 and a network of hydrogen bonds

a α1-D γ2-C
b c
Mb38 β3-E
Loop-C 35
Loop-C
α1-A
30
Distance along pore (Å)

Y157
E155 25
β3-B Y97
Y205 F65 20
β6 9′
GABA
T130
β3-E F200 15 PTX 6′
β2 10
90° 2′ PTX
T202
TMD

Loop-C 5 PTX/
γ2-C PTX –2′ GABA
R67
β3-E+ α1-D– β1 0
α1-D 0 1 2 3 4 5
Pore radius (Å)
PIP2

d e α1 helix f α1 helix
GABA
1.3° β3-B+ α1-A– β3-E+ α1-D–

2.6° ECD rotation F15


F31

γ2-C F15 β3 β5 F31 β3


β3-B β5
Y159 Y159
R85 β6
D163 β6 D163
R85
α1-D
α1-A 0.9° R120 R120
β2
β3-E
β2 Loop-B
Loop-B

β1
1.3° Loop-C Y205 GABA
Y205 GABA
β1
PTX
Loop-C Loop-C
PTX/GABA 4.5°

Fig. 2 | Conformational impact of GABA binding to the α1β3γ2L structures based on the global TMD alignment. Subunits were radially
GABAA receptor. a, Cryo-EM map of the PTX/GABA-bound α1β3γ2L translated away by 10 Å from the pore axis to enable better visualization of
receptor viewed from the extracellular space (left) and parallel to the conformational changes in the ECD upon GABA binding. GABA-induced
membrane plane (right). b, One GABA-binding pocket viewed from the ECD rotation is stated as the rotation angle around the ECD rotation axes,
extracellular space. GABA is shown in ball-and-stick representation with and the direction of motion is shown. e, f, Superposition of α1 ECDs from
the atoms coloured as follows: carbon, khaki; oxygen, red; nitrogen, blue. PTX-bound and PTX/GABA-bound structures reveals conformational
c, Plot of the pore radii for the receptor bound to PTX (black dashed changes induced by GABA binding in the orthosteric pockets at β3-B+/
line) and PTX/GABA (green line). d, Superposition of ECDs from the α1-A− (e) and β3-E+/α1-D− (f) interfaces. Dashed lines indicate hydrogen
PTX-bound (grey) and the PTX/GABA-bound (red/blue/yellow) receptor bonds, π–π stacking, π–cation interactions and salt bridges.

N A T U R E | www.nature.com/nature
© 2018 Springer Nature Limited. All rights reserved.
Article RESEARCH

a 3 μM Mb38
b 3 μM Mb38 100 μM BCC c 3 μM Mb38
of the extracellular domain (ECD) and consequently modulates agonist
binding. Indeed, PTX has been shown to reduce the apparent agonist
affinity in GABAA receptors26,36 and to increase GABA dissociation37.
Therefore, TMD closure by PTX constitutes the basis for allosteric com-
munication between the PTX and GABA binding sites and explains
how PTX appears to act as a competitive inhibitor34,38,39 without
binding to other receptor pockets. We conclude that the PTX-bound
200 pA
10 s structure best illustrates the α1β3γ2L receptor in a closed/resting state
d (closed TMD, agonist-free ECD).
α1-A β3-E α1-D γ2-C
β3-E
β3-B
Mechanistic impact of GABA binding
Previous observations suggest that the two GABA-binding sites
Loop-C
may not be functionally equivalent21,40, and the β-E+/α-D− site has
been estimated to have a threefold higher affinity for GABA than
BCC
the β-B+/α-A− site21. By comparing ECDs in the PTX-bound and
Mb38
the PTX/GABA-bound structures, both of which have closed state
Out TMDs, it is possible to visualize the initial conformational changes at
the two agonist-binding sites upon the binding of GABA. Closure of β3
140° loops-C—likely to be the first step in the process—triggers remodelling
of the β3-B+/α1-A− and the β3-E+/α1-D− interfaces, which leads to
an anticlockwise (looking down the pore axis from the extracellular
In space) asymmetric rotation of all subunit ECDs (Fig. 2d). This state is
PIP2 stabilized by new hydrogen-bond networks involving the β3Gly158,
e f β3Tyr205 and α1Arg85 residues. The cation–π interaction between
35 α1Arg85 and β3Tyr159 breaks, and the α1Arg85 side chain moves to
β5
30 the periphery of the interface (Fig. 2e, f). At the β3-B+/α1-A− inter-
Loop-B
Distance along pore (Å)

β6 face, these changes enable the β3-B subunit to move closer to the α1-A
25
Y205
Y157 subunit by 1.5 Å (as measured between β3Gly33Cα and α1Pro80Cα),
R67
β2 20 9′ and the total buried surface between the two subunit ECDs increases
15 by 184 Å2 (Extended Data Fig. 2j, Extended Data Table 2). In the
Loop-C GABA-bound conformation, α1Phe15 and β3Phe31 residues form a
10 2′ PTX hydrogen–π interaction and the side chains of α1Arg85 and β3Asp163
β1
F200 5 –2′
BCC form new salt bridges, further interlocking the β3-E+/α1-D− ECD
interface and increasing its total buried surface area by 234 Å2 (Fig. 2f,
0
β3-B+
F46
α1-A– 0 1 2 3 4 5 Extended Data Table 2, Supplementary Video 5).
Pore radius (Å) The ‘incomplete’ closure of the β3-B+/α1-A− interface in the PTX/
Fig. 3 | Structure of a α1β3γ2L GABAA receptor closed by the GABA-bound structure, relative to the β3-E+/α1-D− interface, defines
competitive antagonist bicuculline. a–c, Representative whole-cell an intermediate conformation from which GABA can presumably
current traces elicited from the same cell (n = 4) by a 42-s pulse of: Mb38 dissociate more readily. Notably, in the desensitized (high affinity to
alone (a); Mb38 and BCC co-applied for 13 s at the 25-s mark (b); Mb38 GABA41) receptor states—described later in this paper—both β3+/
again, after BCC was washed out (c). d, Cryo-EM map of the α1β3γ2 α1− interfaces close to the same extent. We conclude that the PTX/
GABAA receptor–BCC complex viewed parallel to the membrane plane. GABA-bound structure represents a pre-active receptor state, in which
e, One BCC binding pocket at the β3+/α1− interface, viewed parallel to the
agonist-induced conformational changes at the ECD level are not large
membrane. Dashed lines indicate π–π interactions and hydrogen bonds.
f, Plot of the pore radii for the α1β3γ2 receptor bound to PTX (black enough to perturb the closed/resting TMD conformation42.
dashed line) and BCC (magenta line).
Mechanism of bicuculline antagonism
Being a highly efficient competitive antagonist, BCC was expected
involving the β3Glu155 carboxyl, main-chain carbonyls of β3Ser156 to induce a true GABAA receptor closed state43. We first verified
and β3Tyr157, and the β3Tyr97 hydroxyl. The GABA carboxylate forms whether BCC could close the α1β3γ2L receptor that had been pre-
salt bridges with α1Arg67 and hydrogen bonds with α1Thr130 and bound to Mb38. In whole-cell voltage-clamp experiments, applica-
β3Thr202 (Fig. 2b). The GABA binding modes that we observe are tion of a 42-s pulse of Mb38 produced currents (Fig. 3a) that were
generally consistent with those proposed in lower-resolution α1β1γ2 inhibited (102 ± 7%; n = 4) by the co-application of bicuculline (100
and α1β2γ2 GABAA receptor models18,19; however, there are some dif- μM) (Fig. 3b) in a reversible fashion (Fig. 3c). We solved the cryo-EM
ferences in the details, illustrated in the accompanying Letter29. structure of the α1β3γ2L heteromer in complex with Mb38 and BCC
to a nominal resolution of 3.69 Å (Fig. 3d, Extended Data Fig. 3a–c,
Allosteric cross-talk between PTX and GABA Extended Data Table 1). Electron microscopy densities corresponding
Co-application of PTX (800 μM) with GABA (5 μM) and megabody 38 to BCC were observed at both of the orthosteric agonist sites (Extended
(Mb38; 2 μM; ref. 29) causes rapid and complete inhibition of whole-cell Data Fig. 3d), where the hydrophobic nature of the phthalide
currents in HEK293 cells, with complete recovery after PTX washout and isoquinoline rings of BCC enable its interaction with ‘aromatic
(Extended Data Fig. 2f–h). Unexpectedly, in both PTX-bound and box’ residues β3Tyr157, β3Phe200, β3Tyr205 and α1Phe65 (Fig. 3e,
PTX/GABA-bound structures, the transmembrane domains (TMDs) Supplementary Video 6). Relative to the PTX-bound structure, the
adopt the same conformation—all five M2 helices present the hydro- β3-B and β3-E subunit loops-C flex inward (by around 2.2 Å at the tip;
phobic 9′ Leu side chains towards the centre of the pore, constricting the Extended Data Fig. 3e) in order to accommodate BCC, but they retain
pore radius to ~1.5 Å (Fig. 2c, Extended Data Fig. 2i). This suggests that overall ‘open’ conformations (Extended Data Fig. 3f). The channel
PTX inhibition is not caused by blocking an open pore, but by inducing pore of the BCC-bound α1β3γ2L receptor is fully closed by the M2
and maintaining a closed pore conformation. It is likely that, by sta- residues at three levels—9ʹ, 2ʹ and −2ʹ (Fig. 3f)—and its subunit con-
bilizing the closed/resting TMD state, PTX affects the conformation formations are almost identical to those of the PTX-bound structure

N A T U R E | www.nature.com/nature
© 2018 Springer Nature Limited. All rights reserved.
RESEARCH Article

a Diazepam b α1-D γ2-C


c α1-D γ2-C d
3 O
β3-E γ2-C–
N β3-E
Loop-C
N
5 1 Y210
6 A 9 β6
S205
7 8 β2
Cl
ALP DZP F100
Alprazolam
4 N Out ALP β1
N N
I Y58
N F77
6 1
H102
7 10
α1-D+ N60
8 9 In
Cl PIP2
e f g h
γ2-C– α1-D– 35
Loop-C GABA + Mb38 + DZP
Y210 M2–M3 loop 30

Distance along pore (Å)


β6 M2 25
S205 M3
N265 20
β2 M286 9′
F100 DZP 15 100 pA
DZP β1 M1 10 s
PTX
10
Y58 ALP/GABA
F77 5 DZP/GABA
–2′
H102
F289 0
α1-D+ β3-E+ M236 0 1 2 3 4 5
N60 Pore radius (Å)

Fig. 4 | Structures of a α1β3γ2L GABAA receptor in desensitized states α1− interface TMD region. g, Plot of the pore radii for the receptor
induced by GABA and alprazolam or GABA and diazepam. bound to PTX (black dashed line), ALP/GABA (blue line) and DZP/
a, Structural formulae of diazepam and alprazolam. Diazepine ring atoms GABA (red dashed line). h, Representative current traces evoked by co-
are numbered. Imidazole (I) and benzene (A) rings are labelled. b, c, The application of GABA (10 mM) with Mb38 (2 μM) and DZP (100 μM) for
cryo-EM map of the α1β3γ2 GABAA receptor in complex with ALP (cyan) 40 s to outside-out patches pulled from HEK293 cells. The currents (n = 6
(b) and DZP (teal) (c) viewed parallel to the membrane plane. d, e, Views patches) desensitized completely in three phases: at slow (0.78 ± 0.84 s−1),
of the benzodiazepine binding site at the α1+/γ2− interface showing ALP medium (16.32 ± 9.34 s−1) and fast (306.5 ± 185.9 s−1) rates. Rate values
(d) and DZP (e) binding modes. Dashed lines indicate π–π interactions are mean ± s.d. The upper black solid bar shows the duration of ligand
and hydrogen bonds. f, The low-affinity DZP-binding site in the β3+/ application.

(Extended Data Fig. 3e, f). The binding of BCC at the orthosteric sites In the DZP/GABA-bound map, we also observed two strong densities
prevents closure of the β3+/α1− subunit interfaces and rotation of the at sites in the β3+/α1− TMD interfaces that are thought to be the binding
ECDs, and also stabilizes TMDs in the closed state, thus inactivating sites of general anaesthethics47 (Extended Data Fig. 4i, Supplementary
the channel. Video 9). These densities are not present in the ALP/GABA-bound map
and have a distinct shape that is attributable to DZP. In these pockets,
Benzodiazepine binding modes and mechanisms the DZP A ring forms hydrophobic interactions with β3Met286 and
We next solved cryo-EM structures of the α1β3γ2L receptor in com- β3Phe289 and the phenyl ring points towards α1Pro233 (Fig. 4f, Extended
plex with GABA and ALP, as well as with GABA and DZP, to nominal Data Fig. 4i). Our structure corroborates previous electrophysiology and
resolutions of 3.26 Å and 3.58 Å, respectively (Fig. 4a–c, Extended Data mutagenesis data, which suggests the existence of secondary DZP-binding
Fig. 4a–f, Extended Data Table 1, Supplementary Video 7). In both sites that are responsible for the anaesthetic activity and biphasic GABAA
structures, GABA molecules are bound to the orthosteric agonist pock- receptor potentiation at higher DZP concentrations40,48.
ets and ALP or DZP occupy the canonical BZD-binding site at the α1+/ We next compared the α1+/γ2− interfaces in PTX-, PTX/GABA-,
γ2− interface, where they form extensive interactions (Fig. 4d, e). The BCC-, ALP/GABA- and DZP/GABA-bound structures. Notably, the
densities of ALP and DZP were well defined, enabling us to distinguish binding of ALP or DZP induced only minor rearrangements in the
the fused benzene-diazepine from the pendant phenyl rings (Extended BZD pocket: γ2Asn60 adopted a different rotamer (Extended Data
Data Fig. 4g, h, Supplementary Video 8). Fig. 5f) and the tip of the α1 loop-C flexed outwards by around 0.6
The binding modes of ALP and DZP shown here are in agreement Å (Extended Data Fig. 5f, g). We therefore propose that BZDs such as
with the results of cysteine crosslinking experiments, in which isothi- ALP and DZP act as ‘connectors’ to stabilize the weakest ECD interface,
ocyanate substitutions at the DZP C7 position reacted with cysteines α1-D+/γ2-C− (Extended Data Table 2), and facilitate the concerted
introduced at the α1His102, α1Asn103 and γ2Asn60 positions, and ECD rotation upon GABA binding. Nanobody 38 (Nb38) and Mb38
isothiocyanate introduced at DZP C3 reacted with the α1Ser206Cys might act similarly by ‘crosslinking’ the α1-A+/β3-E− and α1-D+/
and α1Thr207Cys mutants44. The chlorine atoms at the C8 and C7 γ2-C− interfaces20,29. FLZ antagonizes classical BZDs by competing for
positions in ALP and DZP, respectively, interact with the α1His102 side the same pocket; however, because it interacts largely with the α1-D+
chain. In the α4 and α6 subunits the equivalent positions are occupied face18,20, its binding would not confer the same inter-subunit connec-
by arginine residues, the larger side chains of which would sterically tivity benefits (Extended Data Fig. 5b, c).
clash with ALP and DZP. This could explain why classical BZDs do not The channel-pore radii in the ALP/GABA-bound and the DZP/
act on GABAA receptor subtypes containing α4 or α6 subunits22,45,46. GABA-bound structures are 2.6 Å and 2.3 Å, respectively, at the 9′
The ALP-bound and DZP-bound structures also demonstrate that level, and are further constricted to 1.6 Å and 1.8 Å by the −2′ res-
BZDs with a pendant phenyl ring do not share the binding mode idues (Fig. 4g). Whole-cell voltage-clamp experiments indicate that
reported for the benzodiazepine antagonist FLZ and the partial ago- a prolonged exposure to a combination of GABA (10 mM), Mb38 (2
nist BRZ18,20 (Extended Data Fig. 5a–e). Compared with ALP and DZP, μM) and diazepam (100 μM) leads to the complete desensitization of
both FLZ and BRZ bind deeper into the BZD pocket and higher from the α1β3γ2L receptor (Fig. 4h). Subunit comparison between ALP/
the pocket floor, which is delineated by the side chain of γ2Asn60. This GABA-bound and DZP/GABA-bound structures reveals that they
could be due to additional hydrogen bonds between the hydroxyl group adopt similar conformations (Extended Data Fig. 6); this illustrates a
of γ2Thr142 and either the imidazole nitrogen or the ester carbonyl of desensitized state of the receptor, in which it is bound to the agonist
FLZ and BRZ20 (Extended Data Fig. 5c, e). but the channel is closed only at the −2′ gate (Supplementary Video 7).

N A T U R E | www.nature.com/nature
© 2018 Springer Nature Limited. All rights reserved.
Article RESEARCH

a 6.3° GABA b 5.3° interfaces (Fig. 5b, Extended Data Fig. 7c). These motions are trans-
8.5° ALP mitted through interactions between the β1–β2 and the M2–M3 loops,
γ2-C
whereas the β6–β7 (Cys) loops act as pivot points and the TMD bundles
Loop-C
4.7°
1
rotate as rigid bodies (Extended Data Figs. 7d, 8a–f, Supplementary
α1-D
γ2-C 4 Video 10).
β3-B
β3-B 3 2 4.9° The relative flexibility in the M2–M3 loops appears to regulate the
α1-D 9′ Leu efficiency of the ECD–TMD signal transduction. In α1 and γ2 subu-
α1-A
β3-E
nits, the highly conserved arginine residues at the M2 19′ positions
4.2° interact with and rigidify the M2–M3 loops. However, in β3 subunits,
β3-E
5.2° the strictly conserved Lys279 residues displace the 19′ Arg269 side
α1-A
PTX Loop-C 5.1° 4.1° chains, causing them to rotate and wedge in between the M1 and M2
M2–M3 loop
ALP/GABA
8.9° helices of the neighbouring α1 subunits. This restricts the β3 TMD
c
rotations (Extended Data Fig. 8g–l), possibly dampening signal trans-
Closed/resting
Pull duction. In α1 and γ2 subunits, positions equivalent to β3Lys279 are
Pull
occupied by conserved threonine residues, which do not clash with 19′
γ Rotation γ Arg (Extended Data Fig. 8g–l). The M2–M3 loops of β3 therefore seem
β β to be more flexible than those of α1 and γ2 (Extended Data Fig. 7d,
α
GABA
α Supplementary Video 10). In agreement with these observations,
Lock α1β3γ2L receptors containing β3Lys279Thr mutations are 20-fold
α β
ALP
α β
Lock more sensitive to GABA relative to the wild-type receptors49.
We propose that GABA-induced signalling can be described by a
Rotation ‘lock-and-pull’ mechanism. GABA binding triggers loops-C closure in
d Pull β subunits, initiating rotation of their ECDs, which become ‘locked’ to
GABA the neighbouring α− interfaces. These conformational changes ‘pull’
the other ECDs, and transfer to the TMDs, leading to a concerted anti-
β α α γ γ clockwise rotation. The inclusion of a BZD strengthens the α1+/γ2−
β ECD interface to facilitate these motions (Fig. 5c). In β subunits, signal
V53 H56 I68 transduction to TMDs is modulated by the flexible M2–M3 loops,
P273 P278 P288
whereas the ECD rotations of α and γ subunits couple to the TMDs
3 2 9′ 3 2 L259 3 2 9′ 3 2 L264 3 2 9′ 3 2 L274 more efficiently because their M2–M3 loops are more rigid (Fig. 5d).
A253
–2′ A248 –2′ –2′ P263
Conclusion
The structures presented here illustrate how important pharmacological
Closed/ Desensitized compounds, used broadly in research and in the clinic, interact with a
resting
full-length human heteromeric GABAA receptor to modulate its confor-
Fig. 5 | Conformational differences between closed/resting and mation and function. Picrotoxin must initially bind to an open channel
desensitized states in a GABAA receptor. a, Superposition of ECDs from pore and subsequently stabilizes a closed/resting receptor state, which
PTX (grey) and ALP/GABA (red/blue/yellow) structures based on the explains its simultaneous channel blocker and allosteric antagonist activ-
global TMD alignment. Subunits were radially translated by 10 Å away ities. Bicuculline occupies the agonist-binding sites. However, unlike
from the pore axis to enable better visualization of conformational changes GABA, it cannot drive the rotation of β subunits and therefore stabilizes
in the ECD upon GABA and ALP binding. GABA- and ALP-induced
the closed channel pore. Comparison of agonist-free and GABA-bound
ECD rotation angles around the rotational ECD axes, and the direction of
motion are shown. b, Global TMD alignment for the PTX-bound (grey) structures delineates the molecular mechanism by which neurotrans-
and ALP/GABA-bound (coloured) structures. 9ʹ Leu side chains are shown mitter binding to the β3+/α1− interfaces prompts a global rotation of
as sticks. c, Schematic illustration of conformational changes initiated by ECD regions, initially in an asymmetrical fashion, and explains how
GABA binding at the ECD level. GABA stabilizes closure of loop-C in each different subunit types transduce this conformational change to their
β subunit, causing ECDs to rotate and form stronger β3+/α1− interfaces. TMDs. We also characterize the binding sites of two major classical
The black arrows indicate the direction of rotation, with increasing arrow benzodiazepines—alprazolam and diazepam—and define their pri-
thickness representing a greater magnitude of rotation. BZDs such as mary role as stabilizers of the α+/γ− interface, facilitating the concerted
alprazolam bind at the α1+/γ2− interface and reinforce it, facilitating motion of GABAA receptor subunits. This work underlines the poten-
the concerted rotation of the ECDs. Black bars (‘stitches’) at the subunit tial of cryo-EM to study the interactions of drugs with challenging yet
interfaces represent the strength of the interfaces (see Extended Data
Table 2). d, Differences in the relative orientations of ECD–TMD between
highly valuable human membrane-protein targets50. Specifically, these
the closed/resting and desensitized states illustrate how GABA binding structures might lead to the rational design of safer and more specific
and ECD rotation affect TMDs. Notably, the M2–M3 loops in β subunits anxiolytic, sedative, hypnotic and anticonvulsant drugs.
deform more than the α and γ equivalents, resulting in lower degrees of
M2 tilt and TMD rotation. Online content
Any methods, additional references, Nature Research reporting summaries, source
data, statements of data availability and associated accession codes are available at
Allosteric interactions between ECDs and TMDs https://doi.org/10.1038/s41586-018-0832-5.
To understand how the GABA-induced ECD rotation induces confor- Received: 17 August 2018; Accepted: 23 November 2018;
mational changes in the TMD region, we compared ALP/GABA-bound Published online xx xx xxxx.
and PTX-bound structures (Fig. 5a, b). In the presence of ALP, GABA
binding leads to equal closure of both β3+/α1− interfaces and a larger 1. Sieghart, W. Structure and pharmacology of γ-aminobutyric acidA receptor
ECD rotation compared to the pre-open PTX/GABA-bound structure subtypes. Pharmacol. Rev. 47, 181–234 (1995).
(Fig. 5a, Extended Data Figs. 2j, k, 7a, b, Supplementary Video 10). 2. Barnard, E. A. et al. International Union of Pharmacology. XV. Subtypes of
This conformational change of the ECD triggers an anticlockwise γ-aminobutyric acidA receptors: classification on the basis of subunit structure
and receptor function. Pharmacol. Rev. 50, 291–313 (1998).
rotation of the TMD. The resulting tilt of M2 helices moves the 9′ Leu 3. Sigel, E. & Steinmann, M. E. Structure, function, and modulation of GABAA
side chains away from the channel pore and towards the inter-subunit receptors. J. Biol. Chem. 287, 40224–40231 (2012).

N A T U R E | www.nature.com/nature
© 2018 Springer Nature Limited. All rights reserved.
RESEARCH Article

4. Möhler, H. GABAA receptors in central nervous system disease: anxiety, 38. Mehta, A. K. & Ticku, M. K. Characterization of the picrotoxin site of GABAA
epilepsy, and insomnia. J. Recept. Signal Transduct. 26, 731–740 (2006). receptors. Curr. Protoc. Pharmacol. 8, (2001).
5. Olsen, R. W. Analysis of γ-aminobutyric acid (GABA) type A receptor subtypes 39. Talwar, S. & Lynch, J. W. Phosphorylation mediated structural and functional
using isosteric and allosteric ligands. Neurochem. Res. 39, 1924–1941 (2014). changes in pentameric ligand-gated ion channels: implications for drug
6. Olsen, R. W. Allosteric ligands and their binding sites define γ-aminobutyric acid discovery. Int. J. Biochem. Cell Biol. 53, 218–223 (2014).
(GABA) type A receptor subtypes. Adv. Pharmacol. 73, 167–202 (2015). 40. Olsen, R. W. GABAA receptor: Positive and negative allosteric modulators.
7. Hunkeler, W. Benzodiazepines, the story of the antagonist flumazenil and of the Neuropharmacology 136 (Pt A), 10–22 (2018).
partial agonist bretazenil. Chimia 47, 141–147 (1993). 41. Chang, Y., Ghansah, E., Chen, Y., Ye, J. & Weiss, D. S. Desensitization mechanism
8. Squires, R. F. & Brastrup, C. Benzodiazepine receptors in rat brain. Nature 266, of GABA receptors revealed by single oocyte binding and receptor function.
732–734 (1977). J. Neurosci. 22, 7982–7990 (2002).
9. Sigel, E., Mamalaki, C. & Barnard, E. A. Isolation of a GABA receptor from 42. Gielen, M. & Corringer, P.-J. The dual-gate model for pentameric ligand-gated
bovine brain using a benzodiazepine affinity column. FEBS Lett. 147, 45–48 ion channels activation and desensitization. J. Physiol. (Lond.) 596, 1873–1902
(1982). (2018).
10. Schofield, P. R. et al. Sequence and functional expression of the GABAA receptor 43. Ueno, S., Bracamontes, J., Zorumski, C., Weiss, D. S. & Steinbach, J. H.
shows a ligand-gated receptor super-family. Nature 328, 221–227 (1987). Bicuculline and gabazine are allosteric inhibitors of channel opening of the
11. Hibbs, R. E. & Gouaux, E. Principles of activation and permeation in an GABAA receptor. J. Neurosci. 17, 625–634 (1997).
anion-selective Cys-loop receptor. Nature 474, 54–60 (2011). 44. Middendorp, S. J. et al. Relative positioning of classical benzodiazepines to the
12. Miyazawa, A., Fujiyoshi, Y. & Unwin, N. Structure and gating mechanism of the γ2-subunit of GABAA receptors. ACS Chem. Biol. 9, 1846–1853 (2014).
acetylcholine receptor pore. Nature 423, 949–955 (2003). 45. Wieland, H. A., Lüddens, H. & Seeburg, P. H. A single histidine in GABAA
13. Miller, P. S. & Aricescu, A. R. Crystal structure of a human GABAA receptor. receptors is essential for benzodiazepine agonist binding. J. Biol. Chem. 267,
Nature 512, 270–275 (2014). 1426–1429 (1992).
14. Miller, P. S. et al. Structural basis for GABAA receptor potentiation by 46. Rudolph, U. & Knoflach, F. Beyond classical benzodiazepines: novel therapeutic
neurosteroids. Nat. Struct. Mol. Biol. 24, 986–992 (2017). potential of GABAA receptor subtypes. Nat. Rev. Drug Discov. 10, 685–697
15. Laverty, D. et al. Crystal structures of a GABAA-receptor chimera reveal new (2011).
endogenous neurosteroid-binding sites. Nat. Struct. Mol. Biol. 24, 977–985 47. Forman, S. A. & Miller, K. W. Anesthetic sites and allosteric mechanisms of
(2017). action on Cys-loop ligand-gated ion channels. Can. J. Anaesth. 58, 191–205
16. Chen, Q. et al. Structural basis of neurosteroid anesthetic action on GABAA (2011).
receptors. Nat. Commun. 9, 3972 (2018). 48. Walters, R. J., Hadley, S. H., Morris, K. D. W. & Amin, J. Benzodiazepines act on
17. Liu, S. et al. Cryo-EM structure of the human α5β3 GABAA receptor. Cell Res. 28, GABAA receptors via two distinct and separable mechanisms. Nat. Neurosci. 3,
958–961 (2018). 1274–1281 (2000).
18. Zhu, S. et al. Structure of a human synaptic GABAA receptor. Nature 559, 67–72 49. Fisher, J. L. A lysine residue in the β3 subunit contributes to the regulation of
(2018). GABAA receptor activity by voltage. Mol. Cell. Neurosci. 20, 683–694 (2002).
19. Phulera, S. et al. Cryo-EM structure of the benzodiazepine-sensitive α1β1γ2S 50. Renaud, J.-P. et al. Cryo-EM in drug discovery: achievements, limitations and
tri-heteromeric GABAA receptor in complex with GABA. eLife 7, e39383 prospects. Nat. Rev. Drug Discov. 17, 471–492 (2018).
(2018).
20. Miller, P. et al. Heteromeric GABAA receptor structures in positively-modulated
active states. Preprint at https://www.biorxiv.org/content/ Acknowledgements We thank G. Cannone and S. Chen for cryo-EM assistance
early/2018/06/05/338343 (2018). at the MRC-LMB; Y. Chaban and K. Dent for cryo-EM assistance at the
21. Baumann, S. W., Baur, R. & Sigel, E. Individual properties of the two functional Electron Bio-Imaging Centre (eBIC), Diamond Light Source; J. García-Nafría,
agonist sites in GABAA receptors. J. Neurosci. 23, 11158–11166 (2003). L. Dong, T. Nakane and S. Scheres for advice on cryo-EM data processing;
22. Tan, K. R., Rudolph, U. & Lüscher, C. Hooked on benzodiazepines: GABAA J. Grimmett and T. Darling for IT and computing support; and members of
receptor subtypes and addiction. Trends Neurosci. 34, 188–197 (2011). the Aricescu laboratory for discussions and comments on the manuscript.
23. Knoflach, F. et al. Pharmacological modulation of the diazepam-insensitive This work was supported by the UK Medical Research Council grants MR/
recombinant gamma-aminobutyric acidA receptors alpha 4 beta 2 gamma 2 L009609/1, MC_UP_1201/15 (A.R.A., S.M. and D.L.) and MC_UP_A025_1013
and alpha 6 beta 2 gamma 2. Mol. Pharmacol. 50, 1253–1261 (1996). (J.Z.); UK Biotechnology and Biological Sciences Research Council grant
24. Johnston, G. A. R. Advantages of an antagonist: bicuculline and other GABA BB/M024709/1 (A.R.A. and D.L.); Human Frontier Science Program grant
antagonists. Br. J. Pharmacol. 169, 328–336 (2013). RGP0065/2014 (A.R.A.); Cancer Research UK grant C20724/A14414 (T.M.);
25. Krishek, B. J., Moss, S. J. & Smart, T. G. A functional comparison of the and Swiss National Science Foundation fellowship 168735 (J.Z.). R.D. and
antagonists bicuculline and picrotoxin at recombinant GABAA receptors. K.W.M. were supported by a grant from the National Institute for General
Neuropharmacology 35, 1289–1298 (1996). Medical Sciences (GM 58448) and by the Department of Anesthesia, Critical
26. Goutman, J. D. & Calvo, D. J. Studies on the mechanisms of action of picrotoxin, Care and Pain Medicine, Massachusetts General Hospital. We acknowledge
quercetin and pregnanolone at the GABAρ1 receptor. Br. J. Pharmacol. 141, the support and the use of resources of Instruct-ERIC (PID1271), part of
717–727 (2004). the European Strategy Forum on Research Infrastructures (ESFRI), and the
27. Xu, X.-J., Roberts, D., Zhu, G.-N. & Chang, Y.-C. Competitive antagonists facilitate Research Foundation-Flanders (FWO) for their support of nanobody discovery,
the recovery from desensitization of α1β2γ2 GABAA receptors expressed in and FWO for a doctoral fellowship to T.U.
Xenopus oocytes. Acta Pharmacol. Sin. 37, 1020–1030 (2016).
28. Dostalova, Z. et al. Human α1β3γ2L gamma-aminobutyric acid type A Reviewer information Nature thanks M. Chebib, M. Jansen and H. Nury for their
receptors: high-level production and purification in a functional state. Protein contribution to the peer review of this work.
Sci. 23, 157–166 (2014).
29. Laverty, D. et al. Cryo-EM structure of the human α1β3γ2 GABAA receptor in a Author contributions S.M. and A.R.A. conceived the project. S.M. carried out
lipid bilayer. Nature https://doi.org/10.1038/s41586-018-0833-4 (2019). protein purification, collected and processed the cryo-EM data, and built and
30. Chen, L., Durkin, K. A. & Casida, J. E. Structural model for gamma-aminobutyric refined the models, with assistance from A.R.A. R.D. and K.W.M. designed
acid receptor noncompetitive antagonist binding: widely diverse structures fit and analysed the electrophysiological experiments, which were performed
the same site. Proc. Natl Acad. Sci. USA 103, 5185–5190 (2006). by R.D. T.U., E.P. and J.S. designed and generated Mb38. I.S.M. contributed to
31. Xu, M., Covey, D. F. & Akabas, M. H. Interaction of picrotoxin with GABAA receptor sample screening by cryo-EM. D.L. developed Volta Phase Plate data collection
channel-lining residues probed in cysteine mutants. Biophys. J. 69, 1858–1867 protocols and contributed to model building. D.K. and A.K. contributed to
(1995). cryo-EM data collection on the Krios-S microscope. T.M. contributed to the
32. Gurley, D., Amin, J., Ross, P. C., Weiss, D. S. & White, G. Point mutations in the M2 analysis of structural data. J.Z. developed contrast transfer function refinement
region of the alpha, beta, or gamma subunit of the GABAA channel that abolish algorithms. S.M. and A.R.A. wrote the manuscript with input from all co-authors.
block by picrotoxin. Recept. Channels 3, 13–20 (1995).
33. Bali, M. & Akabas, M. H. The location of a closed channel gate in the GABAA Competing interests The authors declare no competing interests.
receptor channel. J. Gen. Physiol. 129, 145–159 (2007).
34. Wang, D.-S., Mangin, J.-M., Moonen, G., Rigo, J.-M. & Legendre, P. Mechanisms Additional information
for picrotoxin block of α2homomeric glycine receptors. J. Biol. Chem. 281, Extended data is available for this paper at https://doi.org/10.1038/s41586-
3841–3855 (2006). 018-0832-5.
35. Unwin, N. Nicotinic acetylcholine receptor and the structural basis of Supplementary information is available for this paper at https://doi.org/
neuromuscular transmission: insights from Torpedo postsynaptic membranes. 10.1038/s41586-018-0832-5.
Q. Rev. Biophys. 46, 283–322 (2013). Reprints and permissions information is available at http://www.nature.com/
36. Qian, H., Pan, Y., Zhu, Y. & Khalili, P. Picrotoxin accelerates relaxation of GABAC reprints.
receptors. Mol. Pharmacol. 67, 470–479 (2005). Correspondence and requests for materials should be addressed to S.M. or
37. Chang, Y. & Weiss, D. S. Site-specific fluorescence reveals distinct structural K.W.M. or A.R.A.
changes with GABA receptor activation and antagonism. Nat. Neurosci. 5, Publisher’s note: Springer Nature remains neutral with regard to jurisdictional
1163–1168 (2002). claims in published maps and institutional affiliations.

N A T U R E | www.nature.com/nature
© 2018 Springer Nature Limited. All rights reserved.
Article RESEARCH

Methods grids (Quantifoil) for 30 s and then blotted for 5.5 s before plunge-freezing the grids
Data reporting. No statistical methods were used to predetermine sample size. into liquid ethane cooled by liquid nitrogen. Plunge-freezing was performed using
The experiments were not randomized and the investigators were not blinded to a Vitrobot Mark IV (Thermo Fisher Scientific) at approximately 100% humidity
allocation during experiments and outcome assessment. and 14.5 °C.
GABAA receptor production, purification and nanodisc reconstitution. Cryo-EM image collection and processing. Cryo-EM data were collected on a
Human tri-heteromeric α1β3γ2L was expressed in a stable, doxycycline-inducible 300 kV Titan Krios microscopes (Thermo Fisher Scientific) fitted with a GIF-
HEK293S-TetR cell line28 grown in suspension. The cell line was not authenticated Quantum energy filter (Gatan) and Volta Phase Plate (Thermo Fisher Scientific).
or tested for mycoplasma contamination. The receptor comprised the full-length Micrographs were recorded in counting mode using K2 Summit (Gatan) or Falcon
human α1 (UniProtKB P14867), β3 (P28472) and γ2L (P18507-2) subunits, each 3EC (Thermo Fisher Scientific) direct electron detectors. For sample-specific data
under individual antibiotic selection (zeocin, hygromycin-B, geneticin and blastici- collection parameters, see Extended Data Table 2.
din, respectively). For quality control and purification, a Flag tag (DYKDDDDK)51 Datasets were analysed using the same basic processing pipeline in RELION
was fused to the α1 subunit N terminus and a rhodopsin-1D4 tag (TETSQVAPA)52 3.056 as described below. First, MotionCor257 and Gctf58 wrappers in RELION 3.0
was fused to the γ2L subunit C terminus, downstream of a (GGS)3GK spacer were used to motion-correct movies, and to estimate the contrast transfer function
sequence. Cells were grown in FreeStyle 293 expression medium (Gibco) sup- and phase shift parameters, respectively. Poor-quality images were discarded after
plemented with 1% fetal bovine serum (Invitrogen), and antibiotics for selection manual inspection. For particle picking, a Gaussian blob was used as a template to
(geneticin, hygromycin-B, zeocin and blasticidin, Thermo Fisher Scientific) at auto-pick particles from a small set of micrographs and 2D classification was per-
37 °C, 160 r.p.m., 8% CO2. GABAA receptor expression was induced by the addi- formed. Selected 2D classes were used as templates for auto-picking particles from
tion of doxycycline (2 μg ml−1, Sigma) at a cell density of around 2 × 106 cells per all micrographs. Two rounds of reference-free 2D classifications were performed
ml. At the same time, the medium was supplemented with 5 mM sodium butyrate and well-aligned 2D classes showing clear GABAA receptor projections were
and the class I α-mannosidase inhibitor kifunensine (1 mg l−1, Toronto Research selected for 3D reconstruction. An initial reference-free 3D model was generated
Chemicals), in order to boost recombinant protein yields. Owing to this treatment, in RELION 3.0 using stochastic gradient descent (SGD) methodology59. Selected
the majority of N-linked glycans were restricted to the immature, endoplasmic-re- particles were 3D-refined (‘gold standard’ refinement) and Bayesian polishing of
ticulum-type Man9GlcNAc2. Cells were collected by centrifugation around 24 h particles was performed60. Next, the polished particles were classified into eight
after doxycycline addition. All steps of purification were performed at 4 °C or on 3D volumes without particle alignment. Particles from volumes with the highest
ice. Cell pellets from 0.4 l of suspension culture were resuspended by brief vor- resolution were combined and 3D-refined using a soft mask and solvent-flattened
texing in dilution buffer (50 mM HEPES pH 7.6, 300 mM NaCl) supplemented Fourier shell correlations (FSCs). Beam tilt correction and per-particle contrast
with 1% (v/v) mammalian protease inhibitor cocktail (Sigma-Aldrich). Membrane transfer function refinement implementations in RELION 3.0 were applied, result-
proteins were solubilized with 1% (w/v) n-dodecyl β-d-maltopyranoside (DDM, ing in further 0.1–0.2 Å increase in resolution. Notably, for the DZP/GABA-bound
Anatrace; used for BCC- and DZP/GABA-bound samples) or lauryl maltose neo- GABAA receptor dataset, beam tilt correction helped to increase the resolution
pentyl glycol (LMNG, Anatrace; used for PTX-, PTX/GABA-, ALP/GABA-bound from 3.82 Å to 3.58 Å. The resolution was estimated using relion_postprocess with
samples) with cholesterol hemisuccinate (CHS, Anatrace) at a 10:1 molar ratio, the FSC criteria of 0.143. Local map resolution was estimated with MonoRes61.
respectively, for 1 h. Insoluble material was removed by centrifugation (10,000g, The initial atomic model used in this work was obtained from the truncated
30 min). The α1β3γ2 GABAA receptors in the supernatant were captured on 1D4 α1β3γ2 heteromer structure20. First, ECDs and TMDs from this model were fit
affinity resin13 (300 μl) while mixing slowly for 2 h. The beads were recovered by to the PTX/GABA-bound α1β3γ2 heteromer map (3.04 Å nominal resolution)
centrifugation (300g, 5 min) and washed with 100 ml of dilution buffer supple- as rigid bodies using UCSF Chimera62. Coot63 was used to adjust the model and
mented with 0.1% (w/v) DDM:CHS (10:1) or LMNG:CHS (10:1). To reduce the build the regions absent in the truncated receptor form. The model was then sub-
time α1β3γ2 GABAA receptor spends solubilized in detergent, and to streamline jected to several rounds of global refinement and minimization in real space using
protein specimen preparation for cryo-EM, we reconstituted the α1β3γ2 heteromer phenix_real_space_refine64. The resulting model served as a starting point for the
into nanodiscs while it was bound to the 1D4 beads. Receptors were equilibrated other structures, applying the same refinement strategy. The geometry constraint
with 1 ml of dilution buffer containing an excess (40 μl) of a phosphatidylcholine files for small-molecule ligands used in the refinement were generated using the
(POPC, Avanti) and bovine brain lipid (BBL) extract (type I, Folch fraction I, Grade Web Server (Global Phasing). The quality assessment of geometry in all
Sigma-Aldrich) mixture (POPC:BBL = 85:15) for 30 min. POPC and BBL extract models was performed using the MolProbity65 Web Server. For the refinement
stocks (10 and 20 mg ml−1, respectively) were prepared by solubilization in 3% protocol validation, each final model coordinates were displaced by 0.5 Å and
DDM. Beads were collected by centrifugation and an excess of MSP2N2 refined using phenix_real_space_refine against one of each half-map sets pro-
(0.6 mg ml−1 final concentration) was added together with Bio-Beads (40 mg ml−1 duced by RELION 3.0. FSC curves were then calculated between this model and
final concentration) and incubated for 2 h rotating gently. The MSP2N2 belt the half-map used for refinement (‘work’) and the half-map, which was not used
protein was produced as previously described53. After nanodisc reconstitution, the for refinement, (‘free’) using phenix.mtriage66. In addition, the refined models
1D4 resin and Bio-Bead mixture was washed extensively with dilution buffer to were also compared against the final sharpened map (‘full’). For all structures, the
remove empty nanodiscs. For protein elution, 100 μl of mixture containing 1 part separation between FSCwork and FSCfree curves was not significant, indicating that
of dilution buffer and 3 parts of 2 mM 1D4 peptide stock (in MilliQ-grade H2O) the models were not over-refined.
was added onto the 1D4 resin for incubation overnight. The next day, beads were Subunit interface surface and associated free energy parameter analysis was
settled down by a short centrifugation (300g, 5 min) and the eluate was collected. performed using the PDBePISA server67. Pore diameters were calculated using the
Typically, the eluate contained 0.1–0.25 mg ml−1 of α1β3γ2 heteromer, which was HOLE68 plug-in in Coot. Structural figures were prepared using UCSF Chimera62.
then directly used for cryo-EM grid preparation. Global TMD alignments were performed using the align command in PyMol
Mb38 production and purification. Megabodies (Mbs) are chimeric proteins (Schrödinger). TMD boundaries for global alignments are as follows: α1 residues
comprising nanobodies (Nbs) fused to larger scaffold proteins (J.S., manuscript in 233–309 and 392–418, β3 residues 218–302 and 419–447, γ2 residues 233–319 and
preparation). A circular permutant of the extracellular adhesin domain of 411–436. ECD alignments were performed using the match command in UCSF
Helicobacter pylori (HopQ)54 was inserted into the first β-turn connecting β-strands Chimera. ECD boundaries for structural alignments: α1 residues 9–222, β3 res-
A and B of anti-α1 subunit Nb3820 resulting in monomeric MbcHopQ Nb38 (approxi- idues 8–217, γ2 residues 26–232. Root mean square deviations (r.m.s.d.) were
mately 58 kDa). MbcHopQ
Nb38 was expressed as a C-terminally His6- and EPEA-tagged calculated using the r.m.s.d. function in UCSF Chimera. M2–M3 loop boundaries
periplasmic protein in E. coli WK6 cells. Cell cultures were grown at 37 °C (120 for r.m.s.d. calculations are as follows: α1 residues 276–284, β3 residues 271–279,
r.p.m.) in Terrific Broth medium supplemented with ampicillin to an optical den- γ2 residues 286–294. Rotation angles were calculated using UCSF Chimera.
sity of 0.8 at 600 nm followed by induction with 1 mM isopropyl β-d-1-thioga- Electrophysiology. HEK293S cells producing α1β3γ2L receptors28 were grown
lactopyranoside and overnight expression at 28 °C (120 r.p.m.). MbcHopQ Nb38 was on glass coverslips, and expression was induced with 0.1–2 μg ml−1 doxycycline
extracted from periplasm by osmotic shock55, and further purified using nickel for 14 to 28 h depending on the level of current required. For pulling outside-out
affinity chromatography and size-exclusion chromatography (Superdex 200 16/60 macro-patches, we used poly-l-lysine-coated glass coverslips (Becton Dickinson).
column, GE Healthcare) in 10 mM Tris pH 7.3, 140 mM NaCl at 21 °C. Purified Cells were also treated with kifunensine (5 μg ml−1) at the time of induction.
MbcHopQ −1
Nb38 was concentrated to around 15 mg ml , frozen in liquid nitrogen and Currents were recorded in either whole-cell or outside-out configuration of
stored at −80 °C. the patch clamp using an Axopatch 200A amplifier (Molecular Devices). Ligands
Cryo-EM sample preparation. α1β3γ2L heteromeric receptors reconstituted into were applied via a quad-channel super-perfusion pipette coupled to a piezoelectric
nanodiscs were supplemented with small-molecule compounds at the concen- element that switched the super-perfusion solution in <1 ms as described previ-
trations indicated in the main text, and with Mb38 (1–2 μM) to improve particle ously69. Data were acquired using Clampex 8.2 (Molecular Devices). For short
alignment and the proportion of side views in cryo-EM images. In brief, 3.5 μl of drug applications, data were acquired at 10 kHz and filtered at 5 kHz, whereas
sample was applied onto glow-discharged gold R1.2/1.3 300 mesh UltraAuFoil for longer applications, data were collected at 2 kHz and filtered at 1 kHz. Cells or

© 2018 Springer Nature Limited. All rights reserved.


RESEARCH Article

patches were continually perfused with a bath solution consisting of (in mM): 145 51. Einhauer, A. & Jungbauer, A. The FLAG peptide, a versatile fusion tag for the
NaCl, 5 KCl, 10 HEPES, 2 CaCl2, 1 MgCl2 and 10 glucose, pH 7.4 (adjusted with purification of recombinant proteins. J. Biochem. Biophys. Methods 49, 455–465
NaOH). The pipette solution consisted of (in mM): 140 KCl, 10 HEPES, 1 EGTA, 2 (2001).
52. Molday, R. S. & MacKenzie, D. Monoclonal antibodies to rhodopsin:
MgCl2 and 2 Mg-ATP, pH 7.3 (adjusted with KOH). For whole-cell recordings, the characterization, cross-reactivity, and application as structural probes.
open pipette resistances ranged from 1.3–2.5 MΩ, and for outside-out patches they Biochemistry 22, 653–660 (1983).
ranged from 6 to 10 MΩ. For whole-cell recordings, cell capacitance ranged from 53. Ritchie, T. K. et al. Reconstitution of membrane proteins in phospholipid bilayer
5 to 13 pF and series resistance ranged from 0.2 to 2.5 MΩ. Series resistances were nanodiscs. Methods Enzymol. 464, 211–231 (2009).
electronically compensated by >85% with a lag of 10 μs. Cells and patches were 54. Javaheri, A. et al. Helicobacter pylori adhesin HopQ engages in a virulence-
enhancing interaction with human CEACAMs. Nat. Microbiol. 2, 16189 (2016).
voltage-clamped at –52 mV unless otherwise stated. The liquid junction potential 55. Pardon, E. et al. A general protocol for the generation of nanobodies for
of –2 mV between the bath and pipette solution was corrected post-recording. structural biology. Nat. Protoc. 9, 674–693 (2014).
Electrophysiology data analysis. Clampfit 9 (Molecular Devices) was used to 56. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure
measure the peak current amplitudes. Current traces were normalized and pre- determination in RELION-3. eLife 7, e42166 (2018).
pared for presentation in Origin 6 (OriginLab Corporation). All data are pre- 57. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion
for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).
sented as mean ± s.d. Statistical analysis was performed using Prism 6 (GraphPad
58. Zhang, K. Gctf: Real-time CTF determination and correction. J. Struct. Biol. 193,
Software). 1–12 (2016).
Materials. All reagents for electrophysiology solutions were purchased from either 59. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms
Millipore Sigma or Thermo Fisher Scientific. Diazepam, doxycycline and DMSO for rapid unsupervised cryo-EM structure determination. Nat. Methods 14,
were purchased from Millipore Sigma. Bicuculline and kifunensine were purchased 290–296 (2017).
from Tocris. A stock solution of bicuculline (100 mM) was prepared fresh each 60. Zivanov, J., Nakane, T. & Scheres, S. H. W. A Bayesian approach to beam-
induced motion correction in cryo-EM single-particle analysis. IUCrJ https://doi.
day in DMSO. The stock solution of diazepam (500 mM) in DMSO was stored at org/10.1107/S205225251801463X (2019).
−80 °C. A working solution of diazepam (100 μM) in the bath solution was pre- 61. Vilas, J. L. et al. MonoRes: automatic and accurate estimation of local resolution
pared fresh daily and was dissolved by sonication. In control experiments, patches for electron microscopy maps. Structure 26, 337–344 (2018).
exposed to GABA (10 mM) in the presence of DMSO (0.02% used in application 62. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory
of diazepam) did not differ from those exposed to GABA (10 mM) alone. The research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).
63. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of
small Mb38-elicited currents were enhanced by a similar small amount by 0.1% Coot. Acta Crystallogr. D 66, 486–501 (2010).
DMSO (84 ± 41% (n = 3)). 64. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and
Reporting Summary. Further information on research design is available in crystallography. Acta Crystallogr. D 74, 531–544 (2018).
the Nature Research Reporting Summary linked to this paper. 65. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular
crystallography. Acta Crystallogr. D 66, 12–21 (2010).
66. Afonine, P. V. et al. New tools for the analysis and validation of cryo-EM maps
Data availability and atomic models. Acta Crystallogr. D 74, 814–840 (2018).
All data are available from the corresponding authors and/or included in the manu- 67. Krissinel, E. & Henrick, K. Inference of macromolecular assemblies from
script or Supplementary Information. Atomic coordinates for PTX-, PTX/GABA-, crystalline state. J. Mol. Biol. 372, 774–797 (2007).
BCC-, ALP/GABA- and DZP/GABA-bound structures have been deposited in the 68. Smart, O. S., Neduvelil, J. G., Wang, X., Wallace, B. A. & Sansom, M. S. P. HOLE: a
program for the analysis of the pore dimensions of ion channel structural
Protein Data Bank with accession codes 6HUG, 6HUJ, 6HUK, 6HUO and 6HUP,
models. J. Mol. Graph. 14, 354–360, 376 (1996).
respectively, and the cryo-EM density maps have been deposited in the Electron 69. Forman, S. A. A hydrophobic photolabel inhibits nicotinic acetylcholine
Microscopy Data Bank with accession codes EMD-0275, EMD-0279, EMD-0280, receptors via open-channel block following a slow step. Biochemistry 38,
EMD-0282 and EMD-0283, respectively. 14559–14564 (1999).

© 2018 Springer Nature Limited. All rights reserved.


Article RESEARCH

Extended Data Fig. 1 | Single-particle cryo-EM analysis of human and model refined in half-map 1 versus half-map 2 (FSCfree). e, The final,
α1β3γ2L GABAA receptor bound to the channel-blocker picrotoxin. unsharpened cryo-EM map coloured by local resolution (estimated using
a, Representative micrograph of the PTX-bound GABAA receptor particles MonoRes61) shown at a higher contour level (left) and at a lower level
embedded in vitreous ice. b, Representative 2D class averages. c, FSC (middle and right) to highlight the nanodisc belt and flexible intracellular
curves for the 3D reconstruction using gold-standard refinement in domains (ICDs). f, Angular distribution of particle projections. The map
RELION56. Curves are shown for the phase randomization, unmasked, of the GABAA receptor–PTX complex is shown in teal. g, Cryo-EM density
masked and phase-randomization-corrected masked maps. d, Validation segments for the PTX-binding site between residues 2ʹ and 9ʹ of the M2
of model refinement protocol. Curves are shown for model versus summed transmembrane helices.
map (FSCfull), model refined in half-map 1 versus half-map 1 (FSCwork)

© 2018 Springer Nature Limited. All rights reserved.


RESEARCH Article

Extended Data 2 | See next page for caption.

© 2018 Springer Nature Limited. All rights reserved.


Article RESEARCH

Extended Data 2 | Structural and electrophysiological analyses of of GABA (5 μM) plus Mb38 (2 μM), each separated by a 1-min wash:
human α1β3γ2L GABAA receptor in complex with PTX and GABA. control (f); one second after the start of the second 8.8-s pulse, PTX
a, FSC curves for the 3D reconstruction of the GABAA receptor bound (800 μM) was co-applied for 4 s (g); wash control showing full recovery
to PTX and GABA. Curves are shown for the phase randomization, (h). PTX inhibited currents by 106 ± 2.6% (mean ± s.d.; n = 6 cells). In
unmasked, masked and phase-randomization-corrected masked maps. addition, the protocol was repeated with outside-out patches (117 ± 9%
b, Validation of the model refinement protocol. Curves are shown for (mean ± s.d.); n = 5 patches). i, Globally superposed PTX-bound and
model versus summed map (FSCfull), model refined in half-map 1 versus PTX/GABA-bound α1β3γ2 receptor transmembrane domains viewed
half-map 1 (FSCwork) and model refined in half-map 1 versus half-map from the extracellular space. Side chains of 9ʹ Leu residues are shown as
2 (FSCfree). c, The final, unsharpened cryo-EM map coloured by local sticks, whereas PTX is represented as balls and sticks. j, k, Superposition
resolution (estimated using MonoRes61). d, Superposition of the PTX- of α1 subunit ECDs from PTX-bound and PTX/GABA-bound α1β3γ2
bound and PTX/GABA-bound α1β3γ2 receptor based on the global receptors reveal the relative β3 ECD motions towards α1 ECDs, as viewed
TMD alignment. The GABA-induced movements of loop-C in each from outside of the receptor (j) and from the vestibule (k). Differences in
of the β3 subunits are highlighted by green lines between Cα atoms of distances (Å) between the selected Cα atoms in the complexes without and
Thr202 residues. GABA is shown as spheres (carbon, khaki; nitrogen, with GABA are indicated by lines. The PTX-bound structure is shown in
blue; oxygen, red). e, Cryo-EM density segments showing GABA-binding grey and the PTX/GABA-bound structure is coloured by subunit (α1, red;
sites in the PTX/GABA-bound structure. f–h, Representative whole-cell β3, blue; γ2, yellow).
current traces elicited from the same HEK293 cell by three 8.8-s pulses

© 2018 Springer Nature Limited. All rights reserved.


RESEARCH Article

Extended Data Fig. 3 | Structural analysis of human α1β3γ2L GABAA e, Superposition of the PTX-bound and BCC-bound α1β3γ2 receptor
receptor bound to the competitive antagonist bicuculline. a, FSC based on the global TMD alignment. The BCC-induced movements of
curves for the 3D reconstruction of the GABAA receptor bound to BCC. loop-C in each of the β3 subunits are highlighted by green lines between
Curves are shown for the phase randomization, unmasked, masked and Cα atoms of Thr202 residues. BCC is shown as spheres (carbon, khaki;
phase-randomization-corrected masked maps. b, Validation of the model nitrogen, blue; oxygen, red). f, Superposition of individual subunits from
refinement protocol. Curves are shown for model versus summed map the PTX-bound and BCC-bound GABAA receptor structures. r.m.s.d.
(FSCfull), model refined in half-map 1 versus half-map 1 (FSCwork) and values (Å) for equivalent Cα in the entire subunits are shown. Loops-C
model refined in half-map 1 versus half-map 2 (FSCfree). c, The final, are marked by arrows. The PTX-bound structure is shown in grey and the
unsharpened cryo-EM map coloured by local resolution (estimated using BCC-bound structure is coloured by subunit (α1, red; β3, blue; γ2, yellow).
MonoRes61). d, Cryo-EM density maps of the BCC-binding pockets.

© 2018 Springer Nature Limited. All rights reserved.


Article RESEARCH

Extended Data Fig. 4 | Structural analysis of human α1β3γ2L GABAA MonoRes61). d–f, Same as a–c but for the GABAA receptor bound to DZP.
receptor in complexes with alprazolam and diazepam. a, FSC curves g–i, Cryo-EM density maps of the ligand binding sites: ALP binding in
for the 3D reconstruction of the GABAA receptor bound to ALP. Curves the BZD pocket (g), DZP binding in the BZD pocket (h), DZP binding
are shown for the phase randomization, unmasked, masked and phase- in the general-anaesthetic pocket at the β3+/α1− interface (i). Ligands
randomization-corrected masked maps. b, Validation of the model are shown as sticks, with atom colouring as follows: ALP carbons, cyan;
refinement protocol. Curves are shown for model versus summed map DZP carbons, teal; nitrogen, blue; chlorine, green. Side chains of residues
(FSCfull), model refined in half-map 1 versus half-map 1 (FSCwork) and lining the binding pockets are shown as sticks and are numbered. Dotted
model refined in half-map 1 versus half-map 2 (FSCfree). c, The final, circles highlight the difference between the structures of alprazolam and
unsharpened cryo-EM map coloured by local resolution (estimated using diazepam.

© 2018 Springer Nature Limited. All rights reserved.


RESEARCH Article

Extended Data Fig. 5 | The classical benzodiazepines flumazenil the structural alignment shows the relative position of the BRZ15 (white).
and bretazenil bind to the same benzodiazepine pocket in GABAA Grey dashed lines indicate hydrogen bonds that FZL and BRZ form with
receptors, but use different modes. ALP/GABA-bound and DZP/GABA- γ2Thr142 in the BZD pocket. f, Superposition of γ2 subunit ECDs from
bound structures are coloured by subunit (α1, red; β3, blue; γ2, yellow), the PTX-, PTX/GABA-, BCC-, DZP/GABA- to ALP/GABA-bound γ2
whereas the other superposed structures are shown in grey. Loop-C is ECD illustrates conformational changes of the BZD pocket associated with
shown in a coil representation, to enable better visualization of the BZD ALP/DZP binding: an outward movement of loop-C; rearrangement of
pocket. a, Structural formulae of flumazenil (FZL) and (S)-bretazenil γ2Tyr58 and γ2Phe77 side chains; and a change in the γ2Asn60 rotamer.
(BRZ). b, c, Superposition of γ2 subunit ECDs from the DZP-bound g, Superposition of the α1-D subunit ECDs from the PTX-bound and
α1β3γ2 and FZL-bound α1β2γ2 receptor structures reveals the FZL16 ALP/GABA-bound α1β3γ2 structures shows that ALP binding causes
(white) position in the BZD pocket relative to DZP (teal). Side-on (b) only a minimal outwards motion of the α1 loop-C, by 0.8 Å as measured
and top-down (c) views of the pocket are presented. d, e, Same as b, c but between Ser206 Cα atom positions.

© 2018 Springer Nature Limited. All rights reserved.


Article RESEARCH

Extended Data Fig. 6 | Superposition of individual subunits from r.m.s.d. values are in the range of 0.38–0.41 Å for α1 (343–344 equivalent
ALP-bound and DZP-bound α1β3γ2L GABAA receptor structures. Cα positions), 0.37–0.40 Å for β3 (334–336 equivalent Cα positions) and
Superposition of the individual subunits from the DZP-bound (grey) and 0.47 Å for γ2 subunits (330 equivalent Cα positions). Loops-C are marked
ALP-bound (α1, red; β3, blue; γ2, yellow) GABAA receptor structures. by arrows.

© 2018 Springer Nature Limited. All rights reserved.


RESEARCH Article

Extended Data Fig. 7 | Structural analysis of PTX-bound and ALP/ bound and the ALP/GABA-bound GABAA receptor structures superposed
GABA-bound α1β3γ2L GABAA receptor structures. The PTX-bound on the basis of the global TMD alignment (ALP/GABA TMD over PTX
structure is shown in grey and the ALP/GABA-bound structure is TMD). Angles between vectors representing M2 helices and the pore axis
coloured by subunit (α1, red; β3, blue; γ2, yellow). a, b, Superposition of the PTX-bound structure are shown. Side chains of residues at the −2ʹ
of α1 subunit ECDs from PTX-bound and ALP/GABA-bound GABAA and the 9ʹ positions are shown. d, Superposition of TMDs from PTX-
receptors reveals the relative β3 ECD motions towards α1 ECDs, as viewed bound and ALP/GABA-bound structures. r.m.s.d. values (Å) are shown
from outside of the receptor (a) and from the vestibule (b). Differences in for entire TMDs and for the M2–M3 loops (see Methods for boundary
distances (Å) between the selected Cα atoms in the complexes without and definitions).
with GABA are indicated with lines. c, Individual subunits from the PTX-

© 2018 Springer Nature Limited. All rights reserved.


Article RESEARCH

Extended Data Fig. 8 | Conformational differences at the ECD–TMD shown, the Cαs for these residues are represented as spheres and the
interfaces between PTX-bound (closed) and ALP/GABA-bound distances of displacement indicated. The strictly conserved M2–M3 loop
(desensitized) α1β3γ2L GABAA receptor structures. The PTX- proline residue interacting with the β1–β2 loop is shown for each subunit.
bound structure is shown in grey and the ALP/GABA-bound structure β1–β2 loop motions are indicated by curved arrows. g–l, Conformational
is coloured by subunit (α1, red; β3, blue; γ2, yellow). The TMDs of the differences in the kM2–M3 loop and 19ʹ Arg side chain positions between
principal subunits of PTX-bound and ALP/GABA-bound structures were the PTX-bound and the ALP/GABA-bound structures shown for α-A
superposed enabling visualization of relative movements of neighbouring (g, h), g2-C (i, j) and b3-B (k, l) subunits. Neighbouring subunit M1
ECDs and TMDs. a, b, Structural rearrangements of the ECD–TMD and M2 helices are shown as cylinders. Dashed lines indicate putative
interface between α1-A and β3-E subunits. c, d, Same as a, b, but for γ2-C hydrogen bond interactions between amino acid side chains and main-
and β3-B subunits. e, f, Same as a, b, but for β3-B and α1-A subunits. chain carbonyls.
Amino acid residues present in the β1–β2 loop tip in each subunit are

© 2018 Springer Nature Limited. All rights reserved.


RESEARCH Article

Extended Data Table 1 | Cryo-EM data collection, refinement and validation statistics

*Local resolution range.


Ψ
Resolution at which the FSC between map and model is 0.5.

© 2018 Springer Nature Limited. All rights reserved.


Article RESEARCH

Extended Data Table 2 | Analysis of interfaces between α1β3γ2L GABAA receptor subunits

*Buried surface area per interface (sum of monomer areas buried at the interface, divided by 2, calculated using PDBePISA67).
† i
Δ G (solvation energy gain at complex formation) is the change of the solvation energy of a subunit due to interface formation, in kcal mol−1, calculated using PDBePISA67).

© 2018 Springer Nature Limited. All rights reserved.


nature research | reporting summary
Corresponding author(s): A. Radu Aricescu

Reporting Summary
Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Research policies, see Authors & Referees and the Editorial Policy Checklist.

Statistical parameters
When statistical analyses are reported, confirm that the following items are present in the relevant location (e.g. figure legend, table legend, main
text, or Methods section).
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
An indication of whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.

A description of all covariates tested


A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistics including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient) AND
variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)

For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.

For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated

Clearly defined error bars


State explicitly what error bars represent (e.g. SD, SE, CI)

Our web collection on statistics for biologists may be useful.

Software and code


Policy information about availability of computer code
Data collection FEI EPU, Clampex 8.2

Data analysis MotionCor2, Gctf-v.1.18, RELION 2.1, RELION 3.0, MonoRes, Scipion, UCSF Chimera v1.12, Pymol v2.0.7, Coot 0.8.9, Phenix 1.13,
MolProbity, PDBePISA, HOLE, Clampfit 9.0., Graphpad Prism 6, Igor Pro, Origin 6.
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors/reviewers
upon request. We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information.

Data
April 2018

Policy information about availability of data


All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
- A list of figures that have associated raw data
- A description of any restrictions on data availability
Atomic coordinates of all protein models were deposited in the Protein Data Bank. Cryo-EM density maps were deposited in the Electron Microscopy Data Bank.

1
nature research | reporting summary
Field-specific reporting
Please select the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/authors/policies/ReportingSummary-flat.pdf

Life sciences study design


All studies must disclose on these points even when the disclosure is negative.
Sample size Sample sizes were estimated on the basis of previous studies using similar methods and analyses that are widely published.

Data exclusions A small number of the acquired cryo-EM movies were discarded owing to poor ice, excessive movement or defocus.

Replication All attempts to replicate data were successful.

Randomization Randomization is not relevant to this study.

Blinding Blinding is not relevant to this study.

Reporting for specific materials, systems and methods

Materials & experimental systems Methods


n/a Involved in the study n/a Involved in the study
Unique biological materials ChIP-seq
Antibodies Flow cytometry
Eukaryotic cell lines MRI-based neuroimaging
Palaeontology
Animals and other organisms
Human research participants

Unique biological materials


Policy information about availability of materials
Obtaining unique materials No restrictions.

Antibodies
Antibodies used Rho 1D4 antibody was purchased from the University of British Columbia. The Mb38 megabody is available upon request.

Validation The nanobody Nb38, used to design the megabody Mb38 as described in methods, was validated by surface plasmon resonance
and published elsewhere (doi: https://doi.org/10.1101/338343).

Eukaryotic cell lines


Policy information about cell lines
April 2018

Cell line source(s) The cell line (based on ATCC CRL-3022) expressing the human alpha1beta3gamma2L GABAA receptor has been previously
described (PMID 24288268).

Authentication Authentication was not performed for this study.

Mycoplasma contamination Mycoplasma testing was not performed for this study.

2
Commonly misidentified lines N/A
(See ICLAC register)

nature research | reporting summary


April 2018

Das könnte Ihnen auch gefallen